Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
2.
Mar Drugs ; 21(3)2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36976210

RESUMEN

New bronchopulmonary dysplasia (BPD) is a neonatal disease that is theorized to begin in utero and manifests as reduced alveolarization due to inflammation of the lung. Risk factors for new BPD in human infants include intrauterine growth restriction (IUGR), premature birth (PTB) and formula feeding. Using a mouse model, our group recently reported that a paternal history of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increased his offspring's risk of IUGR, PTB, and new BPD. Additionally, formula supplementation of these neonates worsened the severity of pulmonary disease. In a separate study, we reported that a paternal preconception fish oil diet prevented TCDD-driven IUGR and PTB. Not surprisingly, eliminating these two major risk factors for new BPD also significantly reduced development of neonatal lung disease. However, this prior study did not examine the potential mechanism for fish oil's protective effect. Herein, we sought to determine whether a paternal preconception fish oil diet attenuated toxicant-associated lung inflammation, which is an important contributor to the pathogenesis of new BPD. Compared to offspring of standard diet TCDD-exposed males, offspring of TCDD-exposed males provided a fish oil diet prior to conception exhibited a significant reduction in pulmonary expression of multiple pro-inflammatory mediators (Tlr4, Cxcr2, Il-1 alpha). Additionally, neonatal lungs of pups born to fish oil treated fathers exhibited minimal hemorrhaging or edema. Currently, prevention of BPD is largely focused on maternal strategies to improve health (e.g., smoking cessation) or reduce risk of PTB (e.g., progesterone supplementation). Our studies in mice support a role for also targeting paternal factors to improve pregnancy outcomes and child health.


Asunto(s)
Displasia Broncopulmonar , Grasas Insaturadas en la Dieta , Neumonía , Dibenzodioxinas Policloradas , Nacimiento Prematuro , Humanos , Recién Nacido , Masculino , Lactante , Embarazo , Animales , Femenino , Niño , Ratones , Displasia Broncopulmonar/prevención & control , Displasia Broncopulmonar/patología , Modelos Animales de Enfermedad , Pulmón/patología , Aceites de Pescado/farmacología , Padre , Dieta
3.
Mar Drugs ; 20(6)2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35736193

RESUMEN

Epidemiology and animal studies suggest that a paternal history of toxicant exposure contributes to the developmental origins of health and disease. Using a mouse model, our laboratory previously reported that a paternal history of in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increased his offspring's risk of developing necrotizing enterocolitis (NEC). Additionally, our group and others have found that formula supplementation also increases the risk of NEC in both humans and mice. Our murine studies revealed that intervening with a paternal fish oil diet preconception eliminated the TCDD-associated outcomes that are risk factors for NEC (e.g., intrauterine growth restriction, delayed postnatal growth, and preterm birth). However, the efficacy of a paternal fish oil diet in eliminating the risk of disease development in his offspring was not investigated. Herein, reproductive-age male mice exposed to TCDD in utero were weaned to a standard or fish oil diet for one full cycle of spermatogenesis, then mated to age-matched unexposed females. Their offspring were randomized to a strict maternal milk diet or a supplemental formula diet from postnatal days 7-10. Offspring colon contents and intestines were collected to determine the onset of gut dysbiosis and NEC. We found that a paternal fish oil diet preconception reduced his offspring's risk of toxicant-driven NEC, which was associated with a decrease in the relative abundance of the Firmicutes phylum, but an increase in the relative abundance of the Negativicutes class.


Asunto(s)
Grasas Insaturadas en la Dieta , Enterocolitis Necrotizante , Microbioma Gastrointestinal , Dibenzodioxinas Policloradas , Nacimiento Prematuro , Animales , Animales Recién Nacidos , Dieta , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/prevención & control , Femenino , Aceites de Pescado/farmacología , Humanos , Masculino , Ratones
4.
Artículo en Inglés | MEDLINE | ID: mdl-35162279

RESUMEN

Over the years, industrial accidents and military actions have led to unintentional, large-scale, high-dose human exposure to environmental contaminants with endocrine-disrupting action. These historical events, in addition to laboratory studies, suggest that exposure to toxicants such as dioxins and polychlorinated biphenyls negatively impact the reproductive system and likely influence the development of gynecologic diseases. Although high-level exposure to a single toxicant is rare, humans living in industrialized countries are continuously exposed to a complex mixture of manmade and naturally produced endocrine disruptors, including persistent organic pollutants and heavy metals. Since minorities are more likely to live in areas with known environmental contamination; herein, we conducted a literature review to identify potential associations between toxicant exposure and racial disparities in women's health. Evidence within the literature suggests that the body burden of environmental contaminants, especially in combination with inherent genetic variations, likely contributes to previously observed racial disparities in women's health conditions such as breast cancer, endometriosis, polycystic ovarian syndrome, uterine fibroids, and premature birth.


Asunto(s)
Dioxinas , Disruptores Endocrinos , Contaminantes Ambientales , Bifenilos Policlorados , Dioxinas/toxicidad , Disruptores Endocrinos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Sustancias Peligrosas , Humanos , Bifenilos Policlorados/toxicidad , Embarazo , Salud de la Mujer
5.
Toxics ; 10(1)2021 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-35051049

RESUMEN

New bronchopulmonary dysplasia is a developmental lung disease associated with placental dysfunction and impaired alveolarization. Risk factors for new BPD include prematurity, delayed postnatal growth, the dysregulation of epithelial-to-mesenchymal transition (EMT), and parental exposure to toxicants. Our group previously reported that a history of paternal toxicant exposure increased the risk of prematurity and low birth weight in offspring. A history of paternal toxicant exposure also increased the offspring's risk of new BPD and disease severity was increased in offspring who additionally received a supplemental formula diet, which has also been linked to poor lung development. Risk factors associated with new BPD are well-defined, but it is unclear whether the disease can be prevented. Herein, we assessed whether a paternal fish oil diet could attenuate the development of new BPD in the offspring of toxicant exposed mice, with and without neonatal formula feeding. We investigated the impact of a paternal fish oil diet preconception because we previously reported that this intervention reduces the risk of TCDD associated placental dysfunction, prematurity, and low birth weight. We found that a paternal fish oil diet significantly reduced the risk of new BPD in neonatal mice with a history of paternal toxicant exposure regardless of neonatal diet. Furthermore, our evidence suggests that the protective effects of a paternal fish oil diet are mediated in part by the modulation of small molecules involved in EMT.

6.
Front Physiol ; 12: 807685, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35153815

RESUMEN

Women with endometriosis, the growth of endometrial glands and stroma outside the uterus, commonly also exhibit adenomyosis, the growth of endometrial tissues within the uterine muscle. Each disease is associated with functional alterations in the eutopic endometrium frequently leading to pain, reduced fertility, and an increased risk of adverse pregnancy outcomes. Although the precise etiology of either disease is poorly understood, evidence suggests that the presence of endometriosis may be a contributing factor to the subsequent development of adenomyosis as a consequence of an altered, systemic inflammatory response. Herein, we will discuss the potential role of exposure to environmental toxicants with endocrine disrupting capabilities in the pathogenesis of both endometriosis and adenomyosis. Numerous epidemiology and experimental studies support a role for environmental endocrine disrupting chemicals (EDCs) in the development of endometriosis; however, only a few studies have examined the potential relationship between toxicant exposures and the risk of adenomyosis. Nevertheless, since women with endometriosis are also frequently found to have adenomyosis, discussion of EDC exposure and development of each of these diseases is relevant. We will discuss the potential mechanisms by which EDCs may act to promote the co-development of endometriosis and adenomyosis. Understanding the disease-promoting mechanisms of environmental toxicants related to endometriosis and adenomyosis is paramount to designing more effective treatment(s) and preventative strategies.

7.
Hum Reprod ; 34(4): 702-714, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30789661

RESUMEN

STUDY QUESTION: Does the uterine vasculature play a localized role in promoting stromal cell decidualization in the human endometrium? SUMMARY ANSWER: Our study demonstrated that hemodynamic forces induced secretion of specific endothelial cell-derived prostanoids that enhanced endometrial perivascular decidualization via a paracrine mechanism. WHAT IS KNOWN ALREADY: Differentiation of stromal cell fibroblasts into the specialized decidua of the placenta is a progesterone-dependent process; however, histologically, it has long been noted that the first morphological signs of decidualization appear in the perivascular stroma. These observations suggest that the human endometrial vasculature plays an active role in promoting stromal differentiation. STUDY DESIGN, SIZE, DURATION: Primary human endometrial stromal cells were co-cultured for 14 days with primary uterine microvascular endothelial cells within a microfluidic Organ-on-Chip model of the endometrium. PARTICIPANTS/MATERIALS, SETTING, METHODS: Cultures were maintained with estradiol and a progestin, with or without continuous laminar perfusion to mimic hemodynamic forces derived from the blood flow. Some cultures additionally received exogenous agonist-mediated challenges. Decidualization in the microfluidic model was assessed morphologically and biochemically. ELISA was used to examine the culture effluent for expression of decidualization markers and prostaglandins. Immunofluorescence was used to monitor cyclooxygenase-2 expression in association with decidualization. MAIN RESULTS AND THE ROLE OF CHANCE: A significantly enhanced stromal decidualization response was observed in the co-cultures when the endothelial cells were stimulated with hemodynamic forces (e.g. laminar shear stress) derived from controlled microfluidic perfusion (<0.001). Furthermore, the enhanced progestin-driven stromal differentiation was mediated via cyclooxygenase-2 and the paracrine action of prostaglandin E2 and prostacyclin. Altogether, these translational findings indicate that the vascular endothelium plays a key physiologic role during the early events of perivascular decidualization in the human endometrium. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This report is largely an in vitro study. Although we were able to experimentally mimic hemodynamic forces in our microfluidic model, we have not yet determined the contribution of additional cell types to the decidualization process or determined the precise physiological rates of shear stress that the microvasculature of the endometrium undergoes in vivo. WIDER IMPLICATIONS OF THE FINDINGS: Identification of specific endothelial-derived prostaglandins and their role during endometrial reproductive processes may have clinical utility as therapeutic targets for reproductive disorders such as infertility, endometriosis, adenomyosis, pre-eclampsia and poor pregnancy outcomes. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Veterans Affairs (I01 BX002853), the Bill and Melinda Gates Foundation Grand Challenges Exploration (OPP1159411), the Environmental Toxicology Training Grant (NIH T32 ES007028) and the Environmental Protection Agency STAR Center Grant (83573601). CONFLICT OF INTEREST: The authors report no conflicts of interest. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Decidua/irrigación sanguínea , Decidua/metabolismo , Dinoprostona/metabolismo , Células Endoteliales/metabolismo , Epoprostenol/metabolismo , Hemodinámica/fisiología , Microfluídica/instrumentación , Adolescente , Adulto , Arteriolas/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Ciclooxigenasa 2/metabolismo , Decidua/citología , Femenino , Fibroblastos/metabolismo , Humanos , Microfluídica/métodos , Persona de Mediana Edad , Comunicación Paracrina/fisiología , Células del Estroma/metabolismo , Adulto Joven
8.
Curr Obstet Gynecol Rep ; 8(3): 103-113, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32953240

RESUMEN

PURPOSE OF REVIEW: Current clinical efforts to predict and prevent preterm birth are primarily focused on the mother and have made minimal progress in improving outcomes. However, recent data indicate that paternal factors can also influence timing of birth. Herein, we will review recent human and murine data examining the contribution of the father to pregnancy outcomes with an emphasis on environmental exposures that can negatively impact fertility and the timing of birth. RECENT FINDINGS: Human epidemiology studies now clearly indicate that a variety of paternal factors (age, race, weight, smoking status) can influence sperm quality, birth timing and, in some studies, offspring health. Utilizing a mouse model, our data have 57demonstrated that developmental exposure to the environmental toxicant TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxin) is associated with a transgenerational reduction in sperm number and quality and an increased risk of preterm birth in an unexposed partner. SUMMARY: Toxicant exposure history can clearly influence sperm quality in men and mice. Murine data further indicate that exposures which negatively affect sperm quality also impair placental function, potentially leading to preterm birth and other adverse outcomes. Of particular concern, these changes have been linked to epigenetic alterations within the male germ cell which can then be transmitted across multiple generations. Since it is not possible to prevent an ancestral toxicant exposure in a human population, identifying lifestyle modifications that can be implemented during the preconception period to improve sperm quality should be explored for the therapeutic potential to reduce the incidence of PTB and its sequelae.

9.
Reprod Sci ; 24(8): 1121-1128, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28322132

RESUMEN

Development of adhesions commonly occurs in association with surgery for endometriosis. Even in the absence of surgery, women with endometriosis appear to be at an enhanced risk of developing adhesions. In the current study, we utilized a chimeric mouse model of experimental endometriosis in order to examine the role of inflammasome activation in the development of postsurgical adhesions. Mice were randomized to receive peritoneal injections of human endometrial tissue fragments or endometrial tissue conditioned media (CM) from women with or without endometriosis 16 hours after ovariectomy and placement of an estradiol-releasing silastic capsule. A subset of mice receiving CM was also treated with interleukin (IL) 1 receptor antagonist (IL-1ra). Our studies demonstrate that peritoneal injection of endometrial tissue fragments near the time of surgery resulted in extensive adhesive disease regardless of tissue origin. However, adhesion scores were significantly higher in mice receiving CM from tissues acquired from patients with endometriosis compared to control tissue CM ( P = .0001). Cytokine bead array analysis of endometrial CM revealed enhanced expression of IL-1ß from patients with endometriosis compared to controls ( P < .01). Finally, the ability of human tissue CM to promote adhesive disease was dramatically reduced in mice cotreated with IL-1ra ( P < .0001). Our data implicate enhanced expression of IL-1ß in women with endometriosis as a potential causal factor in their increased susceptibility of developing postsurgical adhesions. Thus, targeting inflammasome activation may be an effective strategy for the prevention of surgical adhesions in patients with endometriosis.


Asunto(s)
Endometriosis/metabolismo , Endometrio/metabolismo , Inflamasomas/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Adherencias Tisulares/metabolismo , Animales , Modelos Animales de Enfermedad , Endometriosis/patología , Endometrio/patología , Endometrio/trasplante , Femenino , Ratones , Adherencias Tisulares/tratamiento farmacológico
10.
Ann Biomed Eng ; 45(7): 1758-1769, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28108942

RESUMEN

The endometrium is the inner lining of the uterus. Following specific cyclic hormonal stimulation, endometrial stromal fibroblasts (stroma) and vascular endothelial cells exhibit morphological and biochemical changes to support embryo implantation and regulate vascular function, respectively. Herein, we integrated a resin-based porous membrane in a dual chamber microfluidic device in polydimethylsiloxane that allows long term in vitro co-culture of human endometrial stromal and endothelial cells. This transparent, 2-µm porous membrane separates the two chambers, allows for the diffusion of small molecules and enables high resolution bright field and fluorescent imaging. Within our primary human co-culture model of stromal and endothelial cells, we simulated the temporal hormone changes occurring during an idealized 28-day menstrual cycle. We observed the successful differentiation of stroma into functional decidual cells, determined by morphology as well as biochemically as measured by increased production of prolactin. By controlling the microfluidic properties of the device, we additionally found that shear stress forces promoted cytoskeleton alignment and tight junction formation in the endothelial layer. Finally, we demonstrated that the endometrial perivascular stroma model was sustainable for up to 4 weeks, remained sensitive to steroids and is suitable for quantitative biochemical analysis. Future utilization of this device will allow the direct evaluation of paracrine and endocrine crosstalk between these two cell types as well as studies of immunological events associated with normal vs. disease-related endometrial microenvironments.


Asunto(s)
Endometrio/irrigación sanguínea , Endometrio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Membranas Artificiales , Modelos Cardiovasculares , Ingeniería de Tejidos/métodos , Técnicas de Cultivo de Célula , Endometrio/citología , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Porosidad
11.
Reprod Toxicol ; 68: 59-71, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27423904

RESUMEN

Humans and other animals are exposed to a wide array of man-made toxicants, many of which act as endocrine disruptors that exhibit differential effects across the lifespan. In humans, while the impact of adult exposure is known for some compounds, the potential consequences of developmental exposure to endocrine disrupting chemicals (EDCs) is more difficult to ascertain. Animal studies have revealed that exposure to EDCs prior to puberty can lead to adult reproductive disease and dysfunction. Specifically, in adult female mice with an early life exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), we demonstrated a transgenerational occurrence of several reproductive diseases that have been linked to endometriosis in women. Herein, we review the evidence for TCDD-associated development of adult reproductive disease as well as known epigenetic alterations associated with TCDD and/or endometriosis. We will also introduce new "Organ-on-Chip" models which, combined with our established murine model, are expected to further enhance our ability to examine alterations in gene-environment interactions that lead to heritable disease.


Asunto(s)
Disruptores Endocrinos/toxicidad , Exposición a Riesgos Ambientales/efectos adversos , Dibenzodioxinas Policloradas/toxicidad , Reproducción/efectos de los fármacos , Animales , Endometriosis/inducido químicamente , Endometriosis/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Reproducción/genética
12.
Biol Reprod ; 95(4): 73, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535957

RESUMEN

The common environmental toxicant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD or, commonly, dioxin) is a known endocrine disruptor that has been linked to the development of endometriosis in experimental models. Using a murine model, we previously demonstrated that in utero TCDD exposure promotes the transgenerational development of an "endometriosis-like" uterine phenotype consisting of reduced responsiveness to progesterone, subfertility and an increased risk of preterm birth. Since adenomyosis is frequently observed as a comorbidity in women with endometriosis, herein, we sought to determine the incidence of adenomyosis in non-pregnant mice with a history of direct or indirect TCDD exposure. Using histologic assessment and immunohistochemical staining, we analyzed murine uteri for adenomyosis, microvessel density and expression of estrogen receptors alpha and beta (ESR1 and ESR2). Our studies revealed that unexposed control mice did not exhibit adenomyosis while this disease was frequently observed in mice with a history of early life TCDD exposure. A transgenerational impact of developmental TCDD exposure was demonstrated since a subset of mice with only an indirect exposure (F3) also exhibited adenomyosis. Microvessel density within the uterus was significantly higher in all groups of TCDD exposed mice compared to control animals, with density correlated to the severity of disease. Both ESR1 and ESR2 protein exhibited alterations in expression in experimental mice compared to controls. Similar to women with endometriosis, we observed a significant reduction in the ratio of Esr1/Esr2 mRNA in all F1 mice compared to controls. Although this retrospective study was not designed to specifically address mechanisms associated with development of adenomyosis, our data suggest that developmental TCDD exposure permanently alters adult steroid responses which may contribute to the transgenerational development of adenomyosis.

13.
Reprod Sci ; 23(1): 11-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26335175

RESUMEN

Endometriosis is an estrogen (ER)-dependent gynecological disease caused by the growth of endometrial tissue at extrauterine sites. Current endocrine therapies address the estrogenic aspect of disease and offer some relief from pain but are associated with significant side effects. Immune dysfunction is also widely believed to be an underlying contributor to the pathogenesis of this disease. This study evaluated an inhibitor of c-Jun N-terminal kinase, bentamapimod (AS602801), which interrupts immune pathways, in 2 rodent endometriosis models. Treatment of nude mice bearing xenografts biopsied from women with endometriosis (BWE) with 30 mg/kg AS602801 caused 29% regression of lesion. Medroxyprogesterone acetate (MPA) or progesterone (PR) alone did not cause regression of BWE lesions, but combining 10 mg/kg AS602801 with MPA caused 38% lesion regression. In human endometrial organ cultures (from healthy women), treatment with AS602801 or MPA reduced matrix metalloproteinase-3 (MMP-3) release into culture medium. In organ cultures established with BWE, PR or MPA failed to inhibit MMP-3 secretion, whereas AS602801 alone or MPA + AS602801 suppressed MMP-3 production. In an autologous rat endometriosis model, AS602801 caused 48% regression of lesions compared to GnRH antagonist Antide (84%). AS602801 reduced inflammatory cytokines in endometriotic lesions, while levels of cytokines in ipsilateral horns were unaffected. Furthermore, AS602801 enhanced natural killer cell activity, without apparent negative effects on uterus. These results indicate that bentamapimod induced regression of endometriotic lesions in endometriosis rodent animal models without suppressing ER action. c-Jun N-terminal kinase inhibition mediated a comprehensive reduction in cytokine secretion and moreover was able to overcome PR resistance.


Asunto(s)
Benzotiazoles/uso terapéutico , Endometriosis/tratamiento farmacológico , Endometrio/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Pirimidinas/uso terapéutico , Adulto , Animales , Benzotiazoles/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Endometriosis/metabolismo , Endometrio/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 3 de la Matriz/metabolismo , Acetato de Medroxiprogesterona/farmacología , Acetato de Medroxiprogesterona/uso terapéutico , Ratones , Ratones Desnudos , Progesterona/farmacología , Progesterona/uso terapéutico , Pirimidinas/farmacología , Ratas
14.
Proc Natl Acad Sci U S A ; 112(31): 9716-21, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26199416

RESUMEN

Endometriosis is a debilitating, estrogen-dependent, progesterone-resistant, inflammatory gynecological disease of reproductive age women. Two major clinical symptoms of endometriosis are chronic intolerable pelvic pain and subfertility or infertility, which profoundly affect the quality of life in women. Current hormonal therapies to induce a hypoestrogenic state are unsuccessful because of undesirable side effects, reproductive health concerns, and failure to prevent recurrence of disease. There is a fundamental need to identify nonestrogen or nonsteroidal targets for the treatment of endometriosis. Peritoneal fluid concentrations of prostaglandin E2 (PGE2) are higher in women with endometriosis, and this increased PGE2 plays important role in survival and growth of endometriosis lesions. The objective of the present study was to determine the effects of pharmacological inhibition of PGE2 receptors, EP2 and EP4, on molecular and cellular aspects of the pathogenesis of endometriosis and associated clinical symptoms. Using human fluorescent endometriotic cell lines and chimeric mouse model as preclinical testing platform, our results, to our knowledge for the first time, indicate that selective inhibition of EP2/EP4: (i) decreases growth and survival of endometriosis lesions; (ii) decreases angiogenesis and innervation of endometriosis lesions; (iii) suppresses proinflammatory state of dorsal root ganglia neurons to decrease pelvic pain; (iv) decreases proinflammatory, estrogen-dominant, and progesterone-resistant molecular environment of the endometrium and endometriosis lesions; and (v) restores endometrial functional receptivity through multiple mechanisms. Our novel findings provide a molecular and preclinical basis to formulate long-term nonestrogen or nonsteroidal therapy for endometriosis.


Asunto(s)
Endometriosis/tratamiento farmacológico , Endometriosis/patología , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Caspasa 3/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endometrio/irrigación sanguínea , Endometrio/patología , Estrógenos/biosíntesis , Femenino , Humanos , Inflamación/patología , Ratones , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Dolor Pélvico/tratamiento farmacológico , Dolor Pélvico/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroides/uso terapéutico , Xantonas/farmacología , Xantonas/uso terapéutico
15.
J Clin Endocrinol Metab ; 98(3): E455-62, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23386644

RESUMEN

CONTEXT: Growth of endometriotic lesions in rodent model of endometriosis is inhibited by resveratrol, a natural polyphenol with antiproliferative and antiinflammatory properties, and simvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) activity. OBJECTIVE: The objective of the investigation was to study the mechanism of action of resveratrol and its interactions with simvastatin, focusing on cholesterol biosynthesis and HMGCR gene expression and protein activity in primary cultures of human endometrial stromal (HES) cells. METHODS: HES cells were obtained from healthy volunteers. Biosynthesis of cholesterol was assessed by measuring the conversion of [(14)C]acetate to [(14)C]cholesterol. HMGCR mRNA transcripts were quantified by real-time PCR, protein expression by Western blot analysis, and enzyme activity by measuring the conversion of [3-(14)C]3-hydroxy-3-methyl-glutaryl-coenzyme A to [(14)C]mevalonic acid lactone in HES cell microsomes. RESULTS: Resveratrol inhibited cholesterol biosynthesis, HMGCR mRNA, and enzyme activity. Simvastatin inhibited cholesterol biosynthesis and enzyme activity but increased HMGCR mRNA and protein expression. Resveratrol potentiated the inhibitory effects of simvastatin on cholesterol biosynthesis and HMGCR enzyme activity and abrogated the stimulatory effects of simvastatin on HMGCR mRNA transcripts and protein expression. CONCLUSIONS: Resveratrol inhibits key steps of the mevalonate pathway by mechanisms that are partly complementary to and partly comparable with simvastatin via reducing both expression and activity of HMGCR. A combination of resveratrol and simvastatin may be of potential clinical relevance to development new treatments of human endometriosis.


Asunto(s)
Endometrio/citología , Ácido Mevalónico/metabolismo , Simvastatina/farmacología , Estilbenos/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Acetatos/metabolismo , Adulto , Antiinflamatorios no Esteroideos/farmacología , Radioisótopos de Carbono , Colesterol/biosíntesis , Colesterol/metabolismo , Sinergismo Farmacológico , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Cultivo Primario de Células , ARN Mensajero/metabolismo , Resveratrol , Células del Estroma/citología
16.
J Clin Endocrinol Metab ; 98(3): E463-71, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23337719

RESUMEN

CONTEXT: Retinoic acid (RA) may promote survival or apoptosis of cells, depending on the levels of binding proteins: apoptosis-inducing cellular RA binding protein 2 (CRABP2), and cell survival-promoting fatty acid binding protein 5 (FABP5). Increased cellular uptake of retinol and altered actions of RA related to reduced expression of CRABP2 may contribute to the development of endometriosis. Recently statins have been shown to inhibit growth of human endometrial stromal (HES) cells and to reduce the number and size of endometriotic implants in experimental models of this disorder. OBJECTIVE: The objective of the study was to determine whether effects of simvastatin on HES cells and experimental endometriotic implants are related to the modulation of the RA system. METHODS: Effects of simvastatin and RA on proliferation and apoptosis of HES cells were evaluated. Expression of stimulated by RA 6 (STRA6), CRABP2, and FABP5 was determined by real-time PCR and Western blotting. Effects of simvastatin were also evaluated in a nude mouse model of human endometriosis. RESULTS: Simvastatin potentiated an inhibitory effect of RA on growth of HES cells. In HES cells, simvastatin induced expression of STRA6 and CRABP2 but not FABP5. Similarly, simvastatin treatment of nude mice bearing human endometrial xenografts led to an increased expression of CRABP2 and STRA6 proteins in ectopic lesions. CONCLUSIONS: Simvastatin interacts with the RA system, inducing the expression of the key protein regulating the uptake of retinol (STRA6) and the expression of apoptosis-promoting CRABP2. These effects may contribute to cooperative apoptosis-inducing effects of simvastatin and RA and support the examination of these compounds in the treatment of endometriosis.


Asunto(s)
Endometriosis/tratamiento farmacológico , Endometriosis/metabolismo , Endometrio/citología , Simvastatina/farmacología , Células del Estroma/efectos de los fármacos , Tretinoina/metabolismo , Adulto , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quimera , Modelos Animales de Enfermedad , Endometriosis/patología , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Cultivo Primario de Células , Receptores de Ácido Retinoico/genética , Células del Estroma/citología , Células del Estroma/metabolismo
17.
Fertil Steril ; 99(2): 543-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23103017

RESUMEN

OBJECTIVE: To examine whether dietary fish oil supplementation reduces development of spontaneous endometriosis-associated adhesions using an established model. DESIGN: Laboratory-based study. SETTING: Medical center research laboratory. PATIENT(S)/ANIMAL(S): Disease-free women of reproductive age and nude mice. INTERVENTION(S): Women were not provided any intervention. Mice were randomized to receive fish oil supplementation or control diet. MAIN OUTCOME MEASURE(S): Experimental endometriosis was established in mice via injection of human endometrial tissue within 16 hours of ovariectomy. Mice were provided standard or menhaden fish oil-supplemented diets for ≥ 2 weeks before initiation of experimental endometriosis and until killing them 1 week later. At necropsy, mice were examined for the presence and extent of adhesions and endometriotic-like lesions. Tissues were excised and morphologically characterized. RESULT(S): Adhesions/lesions were reduced in mice provided with dietary fish oil compared with control animals. Leukocytes were more numerous within the adhesions/lesions of the mice maintained on the standard diet compared with animals provided with fish oil. As indicated by staining intensity, collagen deposition was greater at adhesion sites within control mice compared with fish oil-supplemented animals. CONCLUSION(S): Wound-healing associated with surgery created an inflammatory peritoneal microenvironment that promoted the development of both experimental endometriosis and adhesions in a murine model. Targeting excessive inflammation with fish oil may be an effective adjuvant therapy to reduce the development of postsurgical adhesions related to endometriosis.


Asunto(s)
Grasas Insaturadas en la Dieta/administración & dosificación , Suplementos Dietéticos , Modelos Animales de Enfermedad , Endometriosis/patología , Endometriosis/prevención & control , Aceites de Pescado/administración & dosificación , Administración Oral , Adulto , Animales , Endometriosis/tratamiento farmacológico , Femenino , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad
18.
Fertil Steril ; 98(4): 948-56.e1, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22789143

RESUMEN

OBJECTIVE: To examine the differentiation-related expression of cannabinoid receptor type 1 (CB1-R) messenger RNA (mRNA) and protein in endometrial tissue obtained from women with and without endometriosis and to determine the impact of acute 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure on CB1-R gene expression in isolated endometrial stromal cells. DESIGN: Laboratory-based study. SETTING: University-affiliated medical center. PATIENT(S): Women with and without endometriosis undergoing volunteer endometrial biopsies after informed consent. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Analysis of in vivo CB1-R mRNA and protein expression in human endometrial tissues and mRNA expression in isolated stromal cells after exposure to TCDD or a progesterone receptor antagonist (onapristone). RESULT(S): Expression of CB1-R mRNA and protein was highest during the progesterone-dominated secretory phase in control samples, but expression was minimal in the endometrial tissues acquired from women with endometriosis, regardless of the cycle phase. Although progesterone was found to induce CB1-R mRNA expression in endometrial stromal cells from control donors, steroid-induced expression of this gene was inhibited by cotreatment with either TCDD or onapristone. CONCLUSION(S): Our studies reveal a role for the anti-inflammatory actions of progesterone in regulating endometrial cannabinoid signaling, which is disrupted in women with endometriosis. We demonstrate for the first time that acute TCDD exposure disrupts cannabinoid signaling in the human endometrium.


Asunto(s)
Endometriosis/fisiopatología , Endometrio/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Progesterona/metabolismo , Receptor Cannabinoide CB1/genética , Adolescente , Adulto , Biopsia , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/patología , Contaminantes Ambientales/toxicidad , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Gonanos/farmacología , Antagonistas de Hormonas/farmacología , Humanos , Interleucina-1alfa/farmacología , Persona de Mediana Edad , Receptor Cannabinoide CB1/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Células del Estroma/fisiología , Adulto Joven
19.
Biol Reprod ; 87(1): 2, 1-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492974

RESUMEN

Recently we reported that statins, the competitive inhibitors of the key enzyme regulating the mevalonate pathway, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR), decrease proliferation of human endometrial stromal (HES) cells. Furthermore, we found that simvastatin treatment reduces the number and the size of endometrial implants in a nude mouse model of endometriosis. The present study was undertaken to investigate the effect of simvastatin on HES cell invasiveness and on expression of selected genes relevant to invasiveness: matrix metalloproteinase 2 (MMP2), MMP3, tissue inhibitor of matrix metalloproteinase 2 (TIMP2), and CD44. Because statin-induced inhibition of HMGCR reduces the production of substrates for isoprenylation-geranylgeranyl pyrophosphate (GGPP) and farnesyl pyrophosphate (FPP)-the effects of GGPP and FPP were also evaluated. Simvastatin induced a concentration-dependent reduction of invasiveness of HES cells. This effect of simvastatin was abrogated by GGPP but not by FPP. Simvastatin also reduced the mRNA levels of MMP2, MMP3, and CD44, but increased TIMP2 mRNA; all these effects of simvastatin were partly or entirely reversed in the presence of GGPP. The present findings provide a novel mechanism of action of simvastatin on endometrial stroma that may explain reduction of endometriosis in animal models of this disease. Furthermore, the presently described effects of simvastatin are likely mediated, at least in part, by inhibition of geranylgeranylation.


Asunto(s)
Endometriosis/tratamiento farmacológico , Endometrio/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simvastatina/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Endometriosis/genética , Endometriosis/metabolismo , Endometriosis/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Fosfatos de Poliisoprenilo/metabolismo , Fosfatos de Poliisoprenilo/farmacología , Prenilación/efectos de los fármacos , Sesquiterpenos/metabolismo , Sesquiterpenos/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/patología , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
20.
Expert Rev Clin Immunol ; 7(5): 611-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21895474

RESUMEN

Endometriosis is a common, complex gynecologic disorder characterized by the presence of endometrial glands and stroma at extrauterine (ectopic) sites. In women who develop this disease, alterations in specific biological processes involving both the endocrine and immune systems have been observed, which may explain the survival and growth of displaced endometrial tissue in affected women. In the past decade, a considerable amount of research has implicated a role for alterations in progesterone action at both eutopic and ectopic sites of endometrial growth which may contribute to the excessive inflammation associated with progression of endometriosis; however, it remains unclear whether these anomalies induce the condition or are simply a consequence of the disease process. In this article, we summarize current knowledge of alterations within the immune system of endometriosis patients and discuss how endometrial cells from women with this disease not only have the capacity to escape immunosurveillance, but also use inflammatory mechanisms to promote their growth within the peritoneal cavity. Finally, we discuss evidence that exposure to an environmental endocrine disruptor, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin, can mediate the development of an endometrial phenotype that exhibits both reduced progesterone responsiveness and hypersensitivity to proinflammatory stimuli mimicking the endometriosis phenotype. Future studies in women with endometriosis should consider whether a heightened inflammatory response within the peritoneal microenvironment contributes to the development and persistence of this disease.


Asunto(s)
Endometriosis/inmunología , Animales , Endometriosis/etiología , Endometriosis/patología , Endometriosis/terapia , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Vigilancia Inmunológica/inmunología , Dibenzodioxinas Policloradas/efectos adversos , Progesterona/inmunología , Teratógenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA